Relapse and non-relapse prediction using a sensitive circulating tumor DNA assay during colorectal cancer postoperative surveillance ==================================================================================================================================== * Tomoko Sasaki * Takeshi Iwaya * Mizunori Yaegashi * Masashi Idogawa * Hayato Hiraki * Masakazu Abe * Yuka Koizumi * Noriyuki Sasaki * Akiko Yashima-Abo * Ryosuke Fujisawa * Fumitaka Endo * Shoichiro Tange * Tomomi Hirano * Koki Otsuka * Akira Sasaki * Mari Masuda * Masashi Fujita * Hidewaki Nakagawa * Fumiaki Takahashi * Yasushi Sasaki * Takashi Tokino * Satoshi S. Nishizuka ## BACKGROUND & AIMS This study investigated whether a circulating tumor DNA (ctDNA) assay using digital PCR (dPCR) could provide early relapse detection and disease-free corroboration at molecular level during postoperative surveillance of colorectal cancer (CRC). **METHODS** The ctDNA dynamics of 52 patients with CRC measured by dPCR targeting 87 individual tumor-specific mutations (1-5 per patient) were compared with results for conventional surveillance using serum tumor markers and computed tomography scanning (CTS). The data were collected between March 2016 and June 2018. **RESULTS** A total of 1,526 prospectively collected plasma samples from 867 timepoints were analyzed. The average number of ctDNA assays per patient was 16.4 and the median follow-up was 1,503 days (range 322-1,951 days). Among patients with Stage II or higher disease who underwent curative resection as their initial surgery (n=47), patients who were ctDNA-positive during the postoperative period (n= 9) showed a higher risk of relapse than those who had sustained ctDNA-negative results (n=38) (hazard ratio 56.3, 95%CI 7.8–407.0, *P* < 0.0001). Elevated ctDNA levels were observed in nine of 10 clinical-relapse patients (11 of 13 events) with an average lead time from a ctDNA-positive time-point to clinical relapse of 191.9 days (range 0-376 days). Given periodic CTS surveillance with ctDNA, 218 (57.1%) CTSs were presumed to be unnecessary for clinical relapse detection and a ctDNA assay would still provide a lead time of 307 days (range 45–582 days). **CONCLUSION** Our findings suggest that the ctDNA assay can reduce the frequency of CTS for relapse diagnosis during postoperative surveillance of CRC. UMIN Clinical Trial Registry number, UMIN000045114 Keywords * circulating tumor DNA * colorectal cancer * postoperative surveillance. digital PCR * tumor-specific mutations In 2020, over 1.8 million new colorectal cancer (CRC) cases and 915,000 deaths were reported worldwide.1 CRC ranks third in incidence, but second in mortality. Approximately two-thirds of patients with stage II or III CRC undergo resection with curative intent.2 Among patients with relapse, resection of liver and lung metastases and systemic chemotherapy have improved patient outcomes.3,4 At present, guidelines established by the National Comprehensive Cancer Network, American Society of Clinical Oncology, European Society for Medical Oncology, and Japanese Society for Cancer of the Colon and Rectum (JSCC) recommend a postoperative surveillance period spanning 5 years during which patients initially diagnosed with stage II or III CRC should be followed up using two types of diagnostic modalities: computed tomography scan (CTS) and serum carcinoembryonic antigen (CEA) level 5–8 The main goal of this surveillance is to improve disease-specific and overall survival (OS) by allowing early detection of relapse and subsequent early treatment intervention. During the 2000s, several meta-analyses demonstrated that intensive follow-up after CRC resection with curative intent resulted in prolonged OS and reduced the re-resection rate for recurrent disease.9–11 However, data from randomized trials and a large cohort study conducted in 2010 and later showed that intensive follow-up for patients with CRC provided no significant benefit.12–15 As such, there is debate about the frequency of surveillance after resection for CRC with curative intent, particularly with respect to the need to balance the potential physical and financial burden associated with intensive surveillance and the advantages of early detection of recurrent disease. Indeed, guidelines still recommend intensive surveillance with CTS every 6-12 months and CEA testing every 3-6 months for patients with CRC during the 5 years after surgery. 5–8 Recently, circulating tumor DNA (ctDNA) has emerged as a promising noninvasive biomarker for the molecular diagnosis and monitoring of several cancer types.16–22 A pan-cancer analysis of ctDNA by Bettegowda et al. demonstrated that ctDNA detection rates were higher for CRC than for other cancer types, with ctDNA being detectable in approximately 70% of patients with localized disease (stages I-III) and 100% of those with metastatic disease (stage IV).23 Clinical validities of the ctDNA assay have been demonstrated for early prediction of therapeutic efficacy in patients with metastatic CRC24–27 and relapse in those with localized CRC.18, 21, 28, 29 A recent analysis showed that reducing adjuvant chemotherapy dose in accordance with ctDNA in stage II CRC patients resulted in substantial cost savings and improved quality-of-life.30 Furthermore, several clinical trials are currently testing the clinical utility of ctDNA testing in CRC patients.31 We previously demonstrated that, in contrast to next-generation sequencing (NGS), frequent ctDNA monitoring by dPCR enabled early relapse prediction, treatment efficacy evaluation, and disease-free corroboration in the management of gastrointestinal cancers, including CRC.32–35 Here, we examined results of ctDNA assays from over 1,500 plasma samples and found that ctDNA monitoring by dPCR can provide early relapse detection and disease-free corroboration at molecular level during postoperative CRC surveillance as compared to conventional CTS and CEA monitoring. ## Materials and Methods ### Patients and sample collection This study was approved by the Institutional Review Board of Iwate Medical University (IRB #HGH28-15 and #MH2021-073). Written informed consent was obtained from all patients. Among 116 patients registered in a clinical study (UMIN Clinical Trial Registry: UMIN000045114) through October 31, 2021, 52 who had undergone complete resection of the tumor and were at least 3 years since the initial surgery were enrolled in this study (Figure 1). These 52 patients were histologically confirmed to have CRC [stages I, II, III, and IV (n=4, 26, 20, and 2, respectively)] and were registered in the study between March 11, 2016, and June 20, 2018. All patients underwent primary tumor resection as a first-line therapy. Nearly all (51/52, 98.1%) of the patients underwent R0 resection at the initial operation, and one patient (CC16010) received a two-stage curative resection for the primary tumor and a metastatic liver tumor. Moreover, 12 patients (1 stage II, 10 stage III, and 1 stage IV) received adjuvant chemotherapy (ACT) after curative resection. A summary of patient characteristics is provided in Supplementary Table 1. Surgically acquired primary tumor tissue samples and corresponding serial blood samples were obtained for the ctDNA assay. In principle, the imaging examinations and blood collection for ctDNA and serum CEA were completed at the same time. ![Figure 1.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2022/09/06/2022.09.03.22279571/F1.medium.gif) [Figure 1.](http://medrxiv.org/content/early/2022/09/06/2022.09.03.22279571/F1) Figure 1. Flow of patient enrollment and sample collection during the study period. CRC, colorectal cancer; ctDNA, circulating tumor DNA; UMIN-CTR, University Hospital Medical Information Network Center Clinical Trials Registry; NGS, next-generation sequencing; PBMC, peripheral blood mononuclear cell; dPCR, digital PCR; CT, computed tomography. ### Panel sequencing of primary tumors DNA samples from the tumor and corresponding peripheral blood mononuclear cells (PBMCs) were subjected to panel sequencing using three different platforms (Supplementary Methods and Figure 1). Briefly, Set 1 was analyzed using the ClearSeq Comprehensive Cancer Panel (Agilent Technologies, Inc., Santa Clara, CA) targeting 151 disease-associated genes on an Illumina Hiseq 2000 sequencer (Illumina, Inc., San Diego, CA).33 Set 2 and Set 3 were analyzed by the Ion Proton™ and the Ion S5™ system (Thermo Fisher Scientific, Waltham, MA), respectively, using a customized panel targeting 39 genes that are frequently altered in CRC (Supplementary Table 2).36 Although the sequencing panel that was used differed among the tumor sets, the major purpose of primary tumor sequencing in this study was to detect a selected number of mutations that have high variant allele frequencies (VAF). All three platforms were sufficient for subsequent ctDNA monitoring in CRC patients. Clonal hematopoiesis of indeterminate potential variants was removed by paired PBMC sequencing for Set 1 and Set 2, and for Set 3, by confirming the absence of the corresponding specific mutation in PBMC DNA by Sanger sequencing. Comprehensive descriptions of methodologies used to identify somatic mutations are presented in the Supplementary Methods.33, 35 ### Monitoring ctDNA levels using dPCR The dPCR assay for quantitative monitoring of ctDNA levels was performed as described previously.32–35 Briefly, specific primers and probes labeled for wild-type and mutant alleles were specifically designed for each mutation identified in a primary tumor, using Hypercool Primer & Probe™ technology (Nihon Gene Research Laboratories, Sendai, Japan). For frequently recurring missense mutations, commercially available primer/probe sets were used (Thermo Fisher Scientific, Waltham, MA and Quantdetect, Inc, Tokyo, Japan). One to five mutations per tumor that had a VAF higher than 10% in primary tumors were prioritized for dPCR analysis. The criteria for mutation selection for the ctDNA assay and the definition of positive and negative ctDNA findings for dPCR are described in the Supplementary Methods. ctDNA data for VAFs were plotted on a time course along with therapy type and clinical information. CEA levels were also measured at the same time points during ctDNA analysis. ### Frequency of CTS during postoperative surveillance According to JSCC guidelines, CTS is recommended at least every 6 months during postoperative surveillance for patients with CRC in Japan. Although the prognostic impact of intensive surveillance for patients with CRC has been controversial, early relapse detection may allow minimally invasive resection of metastatic tumors. From this perspective, at our institute intensive surveillance with CTS concurrent with blood CEA tests every 3-4 months has been performed for early detection of disease recurrence. The number of times patients underwent CTS during the postoperative surveillance period was counted for this cohort. To reduce radiation exposure, side effects associated with contrast medium administration, and medical expense, the number of CTS times should be minimized during the postoperative surveillance period. In this study, the probability of CTS that allowed detection of relapse and delay of relapse detection were evaluated in the context of surveillance and in terms of the reduction in the number of CTS. The number of unnecessary CTS conducted for relapse detection was estimated based on reducing CTS using a designated timing of approximately once per year (i.e., annual CTS) compared to the actual surveillance conducted. CTSs at the closest time points from approximate annual imaging examination day after initial surgery were selected as once-a-year CTS. The delay time was defined as the interval between the actual clinical-relapse detection and the first timepoint when relapse was detected by the designated annual CTS. The delayed first timepoint of CTS was defined as the estimated clinical-relapse timepoint. ### Relapse detection using CTS and ctDNA assay The timing of clinical relapse was defined based on the time point at which a radiologist in daily clinical practice confirmed or suspected the lesion to be a relapse (i.e., the word “relapse” was present in the radiographic report). Meanwhile, ctDNA-relapse was defined as the time of the ctDNA-positive detection of at least two consecutive ctDNA-positive time points. The lead time was defined as days from ctDNA-relapse to clinical-relapse. ### Statistical analysis Diagnostic quality for clinical-relapse by ctDNA and CEA was expressed as sensitivity and specificity, as well as positive/negative predictive values. For group comparisons, the Mann-Whitney U test and Fisher’s exact test were used. Correlations between two variables were calculated based on the Spearman’s rank correlation coefficient. Kaplan-Meier estimates with log-rank tests were used to compare relapse-free survival (RFS), stratified based on the ctDNA status (i.e., positive or negative) before treatment, at the first postoperative time point (post-op), and throughout the postoperative surveillance period. Clinical-RFS based on radiologic finding, estimated clinical-RFS with once-a-year CTS, and ctDNA-RFS based on ctDNA status were also compared. The Cox proportional hazards model was used to estimate risks, based on RFS. We considered *P* values <0.05 to be statistically significant for all analyses. All analyses were performed using GraphPad Prism 8 (GraphPad Software, San Diego, CA), with *P* values <0.05 indicating statistical significance. ## Results ### Mutations in primary CRC At least one somatic mutation was identified in all 52 patients. Supplementary Tables 3 and 4 and Supplementary Figure 1 summarize the mutation profile; the detailed mutation profile for Set 1 is available in our previous report.33 A total of 109 mutations [3 mutations per sample (range, 1-24) on average] and 85 mutations [3 mutations per sample (range, 1-36) on average] were identified using the customized panel for CRC for Sets 2 and 3, respectively. Patient CC16024 in Set 2 and Patient CC16054 in Set 3 had hypermutations with 24 (212.8/Mb) and 36 (319.1/Mb) mutations, respectively. Among the three different sequence platforms, the most frequently mutated genes were *TP53* (37/52, 71.2%), *APC* (28/52, 53.8%), *KRAS* (24/52, 46.2%), *PIK3CA* (13/52, 25.0%), and *BRAF* (8/52, 15.4%). The average VAFs for these five genes were: *TP53*, 44.8% (range, 7.78%-85.9%), *APC*, 33.3% (range, 12.0%-61.0%), *KRAS*, 31.1% (range, 3.6%-81.7%), *PIK3CA*, 28.5% (range, 8.0%-40.5%), and *BRAF*, 28.1% (range, 12.9%-48.2%). Among the 52 patients, 50 (96.2%) had mutations in at least one of the five genes, and 41 of 52 (78.8%) had more than two mutations in one of the five genes. ### Mutations selected for ctDNA detection in CRC patients First, probe/primer sets for the 87 selected mutations from 52 patients were validated by dPCR using corresponding primary tumor DNA (Figure 2A, Supplementary Tables 3 and 4). The number of mutations used per patient for ctDNA testing was: 1, 30 patients; 2, 13 patients; 3, 6 patients; 4, 2 patients and 5, 1 patient (mean 1.7 ± 0.8). With NGS, the mean VAF of the 87 selected mutations in the primary tumor DNA was 35.5% (range, 8.8%–78.3%) and for dPCR the mean VAF was 33.5% (range, 3.6%–85.9%). A strong positive correlation between NGS and dPCR in terms of VAF was observed for the primary tumor DNA (*r* = 0.8, 95% CI, 0.81-1.03, *P* < 0.0001, Spearman’s rank correlation coefficient, Supplementary Figure 2). Fourteen probe/primer sets for mutations identified more than once were used for 36/52 (69.2%) patients (Figure 2A). The percentages of each mutation in all recurrent mutations in this study cohort and that in CRC samples registered in the Catalogue of Somatic Mutation in Cancer (COSMIC) database37 are shown in Figure 2B and C, respectively. The proportion of non-*KRAS/BRAF* mutations in the 48 recurrent mutations of our cohort was higher than that for CRC samples referenced in the COSMIC database [22/48 (45.8%) vs. 2,305/31,379 (7.3%), respectively]. ![Figure 2.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2022/09/06/2022.09.03.22279571/F2.medium.gif) [Figure 2.](http://medrxiv.org/content/early/2022/09/06/2022.09.03.22279571/F2) Figure 2. Somatic mutation profile of primary CRC tumors analyzed by dPCR. (A) Mutation profile of the 52 CRC tumors analyzed by dPCR. Colored boxes indicate the intersection between patients and primer/probe sets. (B) Composition of mutations among recurrent mutations in CRC patients included in the present cohort. (C) Composition of mutations in CRC samples registered in the COSMIC database relative to the recurrent mutations detected in our study. A total of 31,379 mutations were registered in the COSMIC database for the 14 recurrent mutations that were included in the present cohort. VAF, variant allele frequency; NGS, next-generation sequencing; dPCR, digital PCR, COSMIC, Catalogue of somatic mutations in cancer. Preoperative plasma from 31 out of the 52 (59.6%) patients was positive for ctDNA (Figure 3). Of these ctDNA-positive patients, 1/4 (25.0%), 14/26 (53.8%), 15/20 (75.0%), and 1/2 (50%) had stage I, II, III, and IV disease, respectively. No significant differences in ctDNA levels were observed between patients with stage II and stage III disease (*P=*0.60, Mann-Whitney U test) (Supplementary Figure 3). ![Figure 3.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2022/09/06/2022.09.03.22279571/F3.medium.gif) [Figure 3.](http://medrxiv.org/content/early/2022/09/06/2022.09.03.22279571/F3) Figure 3. Longitudinal ctDNA monitoring during the postoperative period for patients with CRC. ctDNA, circulating tumor DNA. Horizontal bars indicate observation periods. ### Longitudinal ctDNA analysis in postoperative surveillance of patients with CRC The median observation period for our cohort was 1,503 (range, 322-1,951) days. A total of 1,526 plasma samples from 867 time points were analyzed for ctDNA. Plasma samples at post-op were collected an average of 34.0 days (range, 20-58) after resection with curative intent. To categorize longitudinal data, ctDNA levels for each time point were binarized (i.e., ctDNA-positive and -negative; Figure 3). Clinical-relapse was observed for 10 out of the 52 (19.2%) patients with CRC. Non-relapsed patients had significantly lower ctDNA-positive rates at all analyzed points compared to relapsed patients [4.0% (27/674) vs. 59.1% (114/193), *P* <0.0001, Fisher’s exact test]. The ctDNA dynamics and detailed clinical information for the 10 relapsed and 42 non-relapsed patients are shown in Figure 4 and Supplementary Figure 4, respectively. The ctDNA dynamics accurately reflected changes in tumor burden by treatment, disease-free status, and clinical-relapse including minimal residual disease. For the 42 patients who had no clinical-relapse, ctDNA-negative results were obtained throughout the postoperative period ranging from 10.7 months to 65.0 months. Of these 42 patients, 23 were pretreatment ctDNA-positive. All exhibited a decrease in ctDNA levels below the detection limit at the post-op time point and continued to be ctDNA-negative. Moreover, 9 out of 10 (90%) patients with clinical relapse showed a decrease in ctDNA in response to treatment and an increase in ctDNA response corresponding to clinical relapse and tumor growth. Taken together, these results indicated that the ctDNA assay provided valid information for tumor burden monitoring during the clinical course for nearly all enrolled patients (51/52; 98.7%). ![Figure 4.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2022/09/06/2022.09.03.22279571/F4.medium.gif) [Figure 4.](http://medrxiv.org/content/early/2022/09/06/2022.09.03.22279571/F4) Figure 4. Dynamics of ctDNA during the postoperative period for CRC patients with relapse. ctDNA, circulating tumor DNA; VAF, variant allele frequency. Among the 10 patients with clinical relapse, Patient CC16010 showed clinical relapse after two-stage resection of the primary tumor and liver metastasis with curative intent (Figure 4). This patient was excluded in the subsequent analysis, because the observation period after liver tumor resection with curative intent was less than three years. Among the 51 patients who underwent resection with curative-intent as the initial surgery, no significant differences in the preoperative ctDNA levels were observed between patients with (n=9) and without clinical relapse (n=42, *P* > 0.05; Mann–Whitney U test). However, those who did have clinical relapse (n=9) showed significantly higher ctDNA VAF than those without clinical relapse at post-op (n=42) (*P* <0.0001, Mann–Whitney U test; Figure 5A and B). ![Figure 5.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2022/09/06/2022.09.03.22279571/F5.medium.gif) [Figure 5.](http://medrxiv.org/content/early/2022/09/06/2022.09.03.22279571/F5) Figure 5. Association between ctDNA status and clinical relapse. (A) Preoperative and (B) postoperative ctDNA levels in CRC patients with or without disease relapse. Plasma samples for the first postoperative time points were collected an average of 34.0 days (range, 20-58) after curative resection. (C)–(E) Relapse-free survival according to ctDNA status at preoperative time point (C), postoperative time point (D), and during the postoperative surveillance period (E). pre-ctDNA, preoperative ctDNA; post-ctDNA, postoperative ctDNA; HR, hazard ratio. *P* values were derived from a Kaplan-Meier log-rank test. HR was calculated using the log-rank test. (F) Frequency of CTS and ctDNA assays during the postoperative surveillance period. Dark and light blue boxes indicate number of CTSs with or without confirmation of clinical-relapse, respectively. Red and pink boxes indicate the number of ctDNA-positive and -negative results, respectively. CTS, computed tomography scan; ctDNA, circulating tumor DNA. The CEA level for each time point was also binarized (i.e., above and below the upper limit of normal) in a swimmer plot (Supplementary Figure 5). Unlike ctDNA, time points at which CEA levels were positive were often observed during the postoperative period in patients without clinical relapse (16/42, 38.1%). Furthermore, for patients with clinical relapse the dynamics of CEA levels did not reflect changes in tumor burden as accurately as ctDNA levels did. No significant difference in CEA level was observed between patients with (n=9) and without clinical-relapse (n=42) at both the pre- and post-op time points (Supplementary Figure 6A and B). The dynamics of ctDNA during the postoperative surveillance period in patients who received ACT but had clinical relapse were associated with ctDNA levels that were continuously or transiently positive (Patients CC16041 and CC16042); during ACT ctDNA elevation (Patients CC16003 and CC16019); and post-ACT ctDNA elevation (Patients CC16011 and CC16030; Figure 4). In patients who had not received ACT, continuous ctDNA-positive (Patient CC16015), transient ctDNA-negative (Patient CC16009), and continuous ctDNA-negative (Patient CC16005) results were obtained. Serial ctDNA analysis during the surveillance period after the initial resection with curative intent in the 51 patients with longitudinal plasma samples identified clinical-relapse with 85.7% sensitivity, 97.6% specificity, 92.3% positive predictive rate (PPR), and 95.4% negative predictive rate (NPR). In contrast, the diagnostic performance of serial CEA analysis for clinical-relapse revealed 64.3% sensitivity, 61.9% specificity, 36.0% PPR, and 83.9% NPR. ### Plasma ctDNA status and risk of recurrence After excluding four patients with stage I disease, we next evaluated the RFS rate stratified with ctDNA status for 47 patients. No significant difference in RFS was observed between preoperative ctDNA-positive (n=28) and -negative (n=19) groups (HR 2.5, 95% CI 0.66–9.35, *P=*0.22, log-rank test; Figure 5C). Patients having sustained post-operative ctDNA-positive results (n=4) showed a significantly higher risk of clinical relapse than those who were ctDNA-negative (n=43) (HR 20.4, 95%CI 0.66-631.0, *P* < 0.0001, log-rank; Figure 5D). Similarly, patients having at least one ctDNA-positive time point during the postoperative surveillance period (n=9) showed a significantly higher risk of clinical relapse than those who had sustained ctDNA-negative results (n=38) (HR 56.3, 95%CI 7.78-407.0, *P* < 0.0001, log-rank test; Figure 5E). Of note, Patient CC16043, who had no clinical relapse, had ctDNA-positive results at the last four time points (Figure 3 and Supplementary Figure 4). Although clinical relapse had not been confirmed as of day 1,703, a suspected lymph node swelling in the abdominal para-aortic area was observed on CTS on day 1,373. In addition, the ctDNA level for the tumor-specific mutation (*TP53* c.G524A) continued to increase from day 1,299 until the last time point (Supplementary Figure 7). ### Possibly unnecessary CTS during postoperative surveillance A total of 382 CTS procedures were performed for our cohort, with an average frequency of 3.3/year per patient. Of these CTS procedures, 13 (3.4%) contributed to confirmation of relapse (including multiple relapses per patient) (Figure 5F). The timing of CTS during the postoperative surveillance period for all patients is presented in Figure 6 and Supplementary Figure 4. A once-a-year interval for CTS during the surveillance period yielded a total of 164 CTS procedures. The rate of CTS that can detect a relapse was significantly increased to 7.9 % (13/164) for once-a-year CTS (Figure 5F). Interestingly, the rate of ctDNA-positive time points out of the total time points was significantly higher than the rate of relapse-confirmed CTSs out of the total number of CTSs carried out (*P* < 0.0001, Fisher’s exact test, Figure 5F). ![Figure 6.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2022/09/06/2022.09.03.22279571/F6.medium.gif) [Figure 6.](http://medrxiv.org/content/early/2022/09/06/2022.09.03.22279571/F6) Figure 6. Lead time for ctDNA elevation and delay in relapse detection during the surveillance period with CTS performed once a year to detect clinical relapse in CRC patients. All time points for CTS and the first time points for ctDNA detection after curative resection during the relapse-free period are shown. ctDNA, circulating tumor DNA; CTS, computed tomography scan; PET, positron emission tomography. From the abovementioned result, among patients with relapse (n=10), CTS might presumably be frequently unnecessary for clinical relapse detection. When the CTS frequency was reduced to once-a-year, delays in clinical relapse detection might have occurred in 7 of the relapsed 10 patients (9 of 13 events), with an average delay of 140.9 days (range, 0-279; Figure 6). In contrast, early ctDNA elevation (ctDNA-relapse) was observed in nine of the 10 relapsed patients (11 of 13 events), with an average lead time from ctDNA-relapse to clinical relapse of 191.9 days (range, 0-376 days; Figure 6). The lead time from ctDNA-relapse to estimated clinical relapse with annual CTS extended to 307.7 days (range, 45-582 days). The clinical-RFS rate for CTS conducted every 3-4 months, estimated clinical-RFS rate with once-a-year CTS, and ctDNA-RFS rate based on ctDNA status were compared for 9 patients who had both clinical- and ctDNA-relapse. No significant difference was observed between the clinical-RFS rate for CTS every 3-4 months and that for once-a-year CTS (HR, 2.0, 95%CI 0.74-5.27, *P=*0.17, log-rank test; Supplementary Figure 8A). In contrast, the clinical-RFS rate with CTS every 3-4 months was significantly higher than the ctDNA-RFS rate (HR, 2.5, 95%CI 0.78-6.78, *P=*0.04, log-rank test; Supplementary Figure 8B). Furthermore, a larger difference was observed between ctDNA-RFS rate and estimated clinical-RFS rate with once-a-year CTS (HR, 3.6, 95%CI 1.16-10.84, *P=*0.0005, log-rank test; Supplementary Figure 8C). ### Relapse events without ctDNA elevation Some relapse events in our study occurred without accompanying ctDNA elevation. In Patient CC16042, a small intrapelvic lesion was pathologically diagnosed as a disseminated relapse after diagnostic surgery (Figure 4 and Supplementary Figure 9). It was suggested that disseminated relapse may have a different DNA fragment-releasing mechanism that resulted in lower ctDNA levels than would be seen for metastatic lesions in other organs.38 Moreover, the tumor showed a low standardized uptake value on positron emission tomography (PET). Thus, one could speculate that tumor cells that have a low cell division rate would release low or undetectable amounts of ctDNA. Indeed, patients with preoperative ctDNA-negative were reported to have substantially higher RFS and OS rates than those who were preoperative ctDNA-positive after hepatectomy for resectable colorectal liver metastases.39 Patient CC16005, who had relapse of liver metastasis, also never had ctDNA-positive results during the surveillance period. Given the lack of clinical- and ctDNA-relapse after resection of liver metastasis over roughly a two-year period, the tumor might comprise dormant cells that have not released ctDNA and in turn the patient would be expected to have a high likelihood of cure after tumor resection. As such, we should consider the possibility that detection of ctDNA might be difficult for some types of tumor relapses. Interestingly, these two cases (CC16042 and CC16005) showed CEA elevation before clinical-relapse diagnosis was made and a decrease after metastatic tumor resection (Supplementary Figure 5). Therefore, the CEA test may be an indispensable tool in postoperative surveillance for CRC patients. Of note, in our 52-patient cohort, one patient (Patient CC16026) showed secondary CRC. In this patient, a positive reaction for the mutation detected in the initial primary cancer (*KRAS* c.G35A) was not observed by dPCR for the primary tumor of the secondary cancer. ## Discussion Several reports have demonstrated that longitudinal ctDNA monitoring during postoperative surveillance could predict clinical-relapse in patients with CRC.18, 21, 29 In such reports, primary tumor sequencing was performed using sequencing panels ranging from targeting of 15 genes to whole-exome sequencing and ctDNA was traced using selected tumor-specific mutations (ranging from 1 to 16 mutations) during surveillance using droplet dPCR or NGS. For the present study, primary tumor DNA was sequenced using three different platforms and then ctDNA was analyzed using a dPCR system with a physical partition (i.e., microwell) on a chip for ctDNA monitoring. Importantly, differences in sequencing platforms appeared to have little impact on the selection of a small number of tumor-specific mutations for ctDNA analysis. The present study analyzed plasma samples from 52 patients across 867 time points, with a median follow-up duration of 50.1 months, which is longer than that for previous studies (12.5-27 months).18, 21,29 Despite the moderate size of the patient cohort (n=52), to our knowledge the number of ctDNA assays per patient was higher than that reported for any previous studies (16.4 vs. 4.6-11.0, respectively) 18, 21, 29. Similar to previous studies,18, 21, 29 patients with ctDNA-positive results at post-op and during the surveillance period also showed significantly higher risk for clinical relapse than those who had ctDNA-negative results using dPCR. The median lead time from ctDNA-relapse to clinical relapse using our dPCR method was 182.0 days (range, 0-376), which is largely consistent with previous studies (5.6 - 11.5 months).18, 21,29 As an extreme case, Patient CC16043 had sustained ctDNA-positive results across over 404 days, but relapse was not clinically confirmed (Supplementary Figure 4). Overall, these results indicate that only a small number of mutations may be sufficient for tumor burden monitoring using a ctDNA assay to predict and detect clinical relapse during postoperative surveillance of CRC patients. With a rate of CTS per million population that is 3-7 times higher compared to that for other industrialized countries, Japan has the highest annual frequency of CT examinations worldwide.40 Patients who undergo cancer resection also have easy access to CTS in Japan. One reason for the difficulty in reducing CTS frequency during disease surveillance may increase the possibility for a delay in relapse detection. In addition, from the perspective of minimally invasive surgery for metastatic tumors, an intensive surveillance with CTS performed every 3-4 months would allow an early detection of recurrence. Consequently, patients included in this study underwent CTS on average 3.3 times/year. In patients with clinical relapse, small lesions could often be retrospectively identified with CTS carried out several months before the clinical relapse diagnosis was made. However, such non-specific findings by CTS did not result in early intervention for relapse treatment. In terms of the timing of clinical relapse diagnosis, no significant difference was observed between the clinical-RFS rate for CTS carried out every 3-4 months compared to that for once-a-year CTS (Supplementary Figure 8A). These results indicated that additional modalities, such as ctDNA monitoring, may be a valuable alternative to increasing the number of CTSs to improve outcomes of postoperative surveillance for CRC. CEA testing has been used for predicting clinical-relapse and could potentially compensate for CTS. In fact, a previous study showed that CRC patients with elevated postoperative CEA had increased risk of recurrence.41 However, our results suggest that serial ctDNA analysis had a higher predictive value for clinical relapse relative to CEA. Unlike ctDNA testing, in terms of longitudinal monitoring CEA testing did not always faithfully reflect changes in tumor burden as evidenced by the high number of false-positive and -negative time points (Figure 3 and Supplementary Figure 5). The lead time from ctDNA-relapse to clinical-relapse of 191.9 days (range, 0-376) for the present cohort indicates that tumor-specific ctDNA detection may indeed be helpful for relapse diagnosis of small lesions that are classified as non-specific findings or cannot be detected with CTS. If CTS is carried out with a longer interval of annual postoperative surveillance, the delay time of clinical-relapse detection will be increased, whereas the timing of ctDNA-relapse will not change. Hence, the difference between “clinical-RFS rate” and “ctDNA-RFS rate” would be larger with less-frequent CTS surveillance. Our findings support a rationale in which CTS for relapse detection conducted at longer intervals would not delay relapse diagnosis if compensatory data is provided by frequent ctDNA assay (e.g., every 3 months). In this cohort, clinical relapse was not observed for 42 patients during the surveillance period. Except for Patient CC16043, who was suspected of having clinical-relapse based on ctDNA testing, all 606 postoperative time points were ctDNA-negative in the other 41 patients who had no clinical relapse. The ability to detect a VAF as low as 0.001% by dPCR42 suggests that a ctDNA-negative result is strongly indicative of disease-free status. As with non-metastatic CRC, Tie et al. recently reported that patients with postoperative and post-adjuvant treatment ctDNA-positive results showed a significantly lower RFS rate than those with ctDNA-negative results after treatment of colorectal liver metastasis.43 Although intensive surveillance may be required for patients who underwent metastatic tumor resection, our personalized, affordable dPCR method can be used to monitor tumor relapse and allow reduced CTS frequency during the postoperative period. Taken together, our results demonstrate that the ctDNA assay may reduce the number of times of CTS is needed during postoperative surveillance of CRC patients without contributing to delays in relapse diagnosis. The current study has some potential limitations. First, our system was not intended to select mutations corresponding to targeted therapies. Second, serial ctDNA monitoring does not detect changes in genetic clonality during therapies. NGS is essential for mutation screening of ctDNA, but not necessarily for monitoring of tumor burden with extremely low VAF ctDNA quantification. Third, in some relapse events occurred in our study, no elevation in ctDNA levels was detected. In conclusion, ctDNA testing can reduce the frequency of CTS from every 3-4 months to at most once-yearly during the postoperative surveillance period in CRC patients without increasing the likelihood of delayed relapse diagnosis. ## Supporting information Supplementary File [[supplements/279571_file08.pdf]](pending:yes) ## Data Availability All data produced in the present work are contained in the manuscript. ## Acknowledgments Author contributions: Takeshi Iwaya, Tomoko Sasaki, Mizunori Yaegashi, and Satoshi S. Nishizuka had full access to all study data. They take responsibility for the integrity of the data and the accuracy of the data analysis. Acquisition, analysis, or interpretation of data: Takeshi Iwaya, Satoshi S. Nishizuka, Tomoko Sasaki, Mizunori Yaegashi, Yuka Koizumi, Akiko Abo, Noriyuki Sasaki, Ryosuke Fujisawa, Fumitaka Endo, Shoichiro Tange, Tomomi Hirano, Masashi Idogawa, Yasushi Sasaki, Masashi Fujita, and Hidewaki Nakagawa. Drafting of the manuscript: Tomoko Sasaki, and Takeshi Iwaya. Critical revision of the manuscript for important intellectual content: Hayato Hiraki, Masakazu Abe, Yasushi Sasaki, Takashi Tokino, and Satoshi S. Nishizuka. Administrative, technical, or material support: Mizunori Yaegashi, Koki Otsuka, Akira Sasaki, Mari Masuda. Study supervision: Satoshi S. Nishizuka, Yasushi Sasaki, Mari Masuda, Fumiaki Takahashi, and Takashi Tokino. We thank the surgeons in Department of Surgery, Iwate Medical University, Drs. Toshimoto Kimura, and Kiyoharu Takashimizu. ## Footnotes * **Conflict of Interest** Dr. Iwaya received grant/research support from Nippon Kayaku, Chugai Pharmaceutical, Daiichi Sankyo and Quantdetect Inc. Dr. Iwaya is a consultant of Quantdetect Inc. Drs. Hiraki and Nishizuka received financial/research support from LSI Medience Co. and Quantdetect Inc. Dr. Hiraki is a consultant of Quantdetect Inc. Dr. Nishizuka received grant/research support form Array Jet, Taiho Pharmaceuticals, Boehringer-Ingelheim, Chugai Pharmaceutical, and Geninus. Dr. Nishizuka is an advisor of CLEA Japan. Dr. Nishizuka is Founder and CEO of Quantdetect Inc. Drs. Iwaya and Nishizuka hold a patent that might benefit from this publication (JP6544783). * **Funding** This work was supported by a Grant-in-Aid for Scientific Research KAKENHI [16K19951, 16K19952, 16H06279, 17K10605, 19K09224, 20K09064, 20K08966, and 21K07223], Keiryokai Collaborative Research Grant [#136 and #145], and Iwate Prefectural Research Grants [H30, R2, and R3] * Received September 3, 2022. * Revision received September 3, 2022. * Accepted September 6, 2022. * © 2022, Posted by Cold Spring Harbor Laboratory The copyright holder for this pre-print is the author. All rights reserved. The material may not be redistributed, re-used or adapted without the author's permission. ## References 1. 1.Sung H, Ferlay J, Siegel RL, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin 2021;71:209–249. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3322/caac.21660&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F09%2F06%2F2022.09.03.22279571.atom) 2. 2.Maringe C, Walters S, Rachet B, et al. Stage at diagnosis and colorectal cancer survival in six high-income countries: a population-based study of patients diagnosed during 2000-2007. Acta Oncol 2013;52:919–32. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3109/0284186X.2013.764008&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23581611&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F09%2F06%2F2022.09.03.22279571.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000318655300006&link_type=ISI) 3. 3.Kanas GP, Taylor A, Primrose JN, et al. Survival after liver resection in metastatic colorectal cancer: review and meta-analysis of prognostic factors. Clin Epidemiol 2012;4:283–301. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.2147/CLEP.S34285&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23152705&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F09%2F06%2F2022.09.03.22279571.atom) 4. 4.Colibaseanu, D. T., Mathis, K. L., Abdelsattar, Z. M., et al. Is curative resection and long-term survival possible for locally re-recurrent colorectal cancer in the pelvis? Dis Colon Rectum 2013;56:9–14 5. 5.National Comprehensive Cancer Network (NCCN). NCCN clinical practice guidelines in oncology. [https://www.nccn.org/professionals/physician_gls](https://www.nccn.org/professionals/physician_gls) 2021. 6. 6.Desch CE, Benson AB, 3rd., Somerfield MR, et al. Colorectal cancer surveillance: 2005 update of an American Society of Clinical Oncology practice guideline. J Clin Oncol 2005;23:8512–9. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamNvIjtzOjU6InJlc2lkIjtzOjEwOiIyMy8zMy84NTEyIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjIvMDkvMDYvMjAyMi4wOS4wMy4yMjI3OTU3MS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 7. 7.Argiles G, Tabernero J, Labianca R, et al. Localised colon cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 2020;31:1291–1305. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.annonc.2020.06.022&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F09%2F06%2F2022.09.03.22279571.atom) 8. 8.Hashiguchi Y, Muro K, Saito Y, et al. Japanese Society for Cancer of the Colon and Rectum (JSCCR) guidelines 2019 for the treatment of colorectal cancer. Int J Clin Oncol 2020;25:1–42. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s10147-019-01485-z&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=31203527&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F09%2F06%2F2022.09.03.22279571.atom) 9. 9.Renehan AG, Egger M, Saunders MP, et al. Mechanisms of improved survival from intensive followup in colorectal cancer: a hypothesis. Br J Cancer 2005;92:430–3. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=15685236&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F09%2F06%2F2022.09.03.22279571.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000226849000004&link_type=ISI) 10. 10.Jeffery M, Hickey BE, Hider PN. Follow-up strategies for patients treated for non-metastatic colorectal cancer. Cochrane Database Syst Rev 2007:CD002200. 11. 11.Tjandra JJ, Chan MK. Follow-up after curative resection of colorectal cancer: a meta-analysis. Dis Colon Rectum 2007;50:1783–99. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s10350-007-9030-5&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=17874269&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F09%2F06%2F2022.09.03.22279571.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000250785500007&link_type=ISI) 12. 12.Primrose JN, Perera R, Gray A, et al. Effect of 3 to 5 years of scheduled CEA and CT follow-up to detect recurrence of colorectal cancer: the FACS randomized clinical trial. JAMA 2014;311:263–70. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1001/jama.2013.285718&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=24430319&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F09%2F06%2F2022.09.03.22279571.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000329566400018&link_type=ISI) 13. 13.Wille-Jorgensen P, Syk I, Smedh K, et al. Effect of More vs Less Frequent Follow-up Testing on Overall and Colorectal Cancer-Specific Mortality in Patients With Stage II or III Colorectal Cancer: The COLOFOL Randomized Clinical Trial. JAMA 2018;319:2095–2103. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F09%2F06%2F2022.09.03.22279571.atom) 14. 14.Rosati G, Ambrosini G, Barni S, et al. A randomized trial of intensive versus minimal surveillance of patients with resected Dukes B2-C colorectal carcinoma. Ann Oncol 2016;27:274–80. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/annonc/mdv541&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26578734&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F09%2F06%2F2022.09.03.22279571.atom) 15. 15.Snyder RA, Hu CY, Cuddy A, et al. Association Between Intensity of Posttreatment Surveillance Testing and Detection of Recurrence in Patients With Colorectal Cancer. JAMA 2018;319:2104–2115. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F09%2F06%2F2022.09.03.22279571.atom) 16. 16.Diehl F, Schmidt K, Choti MA, et al. Circulating mutant DNA to assess tumor dynamics. Nat Med 2008;14:985–90. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nm.1789&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=18670422&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F09%2F06%2F2022.09.03.22279571.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000258988600033&link_type=ISI) 17. 17.Dawson SJ, Tsui DW, Murtaza M, et al. Analysis of circulating tumor DNA to monitor metastatic breast cancer. N Engl J Med 2013;368:1199–209. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa1213261&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23484797&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F09%2F06%2F2022.09.03.22279571.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000316869000007&link_type=ISI) 18. 18.Tie J, Wang Y, Tomasetti C, et al. Circulating tumor DNA analysis detects minimal residual disease and predicts recurrence in patients with stage II colon cancer. Sci Transl Med 2016;8:346ra92. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTE6InNjaXRyYW5zbWVkIjtzOjU6InJlc2lkIjtzOjEzOiI4LzM0Ni8zNDZyYTkyIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjIvMDkvMDYvMjAyMi4wOS4wMy4yMjI3OTU3MS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 19. 19.Tan L, Sandhu S, Lee RJ, et al. Prediction and monitoring of relapse in stage III melanoma using circulating tumor DNA. Ann Oncol 2019;30:804–814. 20. 20.Abbosh C, Birkbak NJ, Wilson GA, et al. Phylogenetic ctDNA analysis depicts early-stage lung cancer evolution. Nature 2017;545:446–451. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nature22364&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=28445469&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F09%2F06%2F2022.09.03.22279571.atom) 21. 21.Reinert T, Henriksen TV, Christensen E, et al. Analysis of Plasma Cell-Free DNA by Ultradeep Sequencing in Patients With Stages I to III Colorectal Cancer. JAMA Oncol 2019;5:1124–1131. 22. 22.Zivanovic Bujak A, Weng CF, Silva MJ, et al. Circulating tumour DNA in metastatic breast cancer to guide clinical trial enrolment and precision oncology: A cohort study. PLoS Med 2020;17:e1003363. 23. 23.Bettegowda C, Sausen M, Leary RJ, et al. Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci Transl Med 2014;6:224ra24. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTE6InNjaXRyYW5zbWVkIjtzOjU6InJlc2lkIjtzOjEzOiI2LzIyNC8yMjRyYTI0IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjIvMDkvMDYvMjAyMi4wOS4wMy4yMjI3OTU3MS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 24. 24.Tie J, Kinde I, Wang Y, et al. Circulating tumor DNA as an early marker of therapeutic response in patients with metastatic colorectal cancer. Ann Oncol 2015;26:1715–22. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/annonc/mdv177&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25851626&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F09%2F06%2F2022.09.03.22279571.atom) 25. 25.Garlan F, Laurent-Puig P, Sefrioui D, et al. Early Evaluation of Circulating Tumor DNA as Marker of Therapeutic Efficacy in Metastatic Colorectal Cancer Patients (PLACOL Study). Clin Cancer Res 2017;23:5416–5425. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6ImNsaW5jYW5yZXMiO3M6NToicmVzaWQiO3M6MTA6IjIzLzE4LzU0MTYiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMi8wOS8wNi8yMDIyLjA5LjAzLjIyMjc5NTcxLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 26. 26.Hsu HC, Lapke N, Wang CW, et al. Targeted Sequencing of Circulating Tumor DNA to Monitor Genetic Variants and Therapeutic Response in Metastatic Colorectal Cancer. Mol Cancer Ther 2018;17:2238–2247. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6Im1vbGNhbnRoZXIiO3M6NToicmVzaWQiO3M6MTA6IjE3LzEwLzIyMzgiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMi8wOS8wNi8yMDIyLjA5LjAzLjIyMjc5NTcxLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 27. 27.Max Ma X, Bendell JC, Hurwitz HI, et al. Disease Monitoring Using Post-induction Circulating Tumor DNA Analysis Following First-Line Therapy in Patients with Metastatic Colorectal Cancer. Clin Cancer Res 2020;26:4010–4017. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6ImNsaW5jYW5yZXMiO3M6NToicmVzaWQiO3M6MTA6IjI2LzE1LzQwMTAiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMi8wOS8wNi8yMDIyLjA5LjAzLjIyMjc5NTcxLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 28. 28.Tie J, Cohen JD, Wang Y, et al. Circulating Tumor DNA Analyses as Markers of Recurrence Risk and Benefit of Adjuvant Therapy for Stage III Colon Cancer. JAMA Oncol 2019;5:1710–1717. 29. 29.Tarazona N, Gimeno-Valiente F, Gambardella V, et al. Targeted next-generation sequencing of circulating-tumor DNA for tracking minimal residual disease in localized colon cancer. Ann Oncol 2019;30:1804–1812. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F09%2F06%2F2022.09.03.22279571.atom) 30. 30.To YH. Circulating Tumour DNA as a Potential Cost-Effective Biomarker to Reduce Adjuvant Chemotherapy Overtreatment in Stage II Colorectal Cancer. Pharmacoeconomics 2021;8:953–964. 31. 31.Dasari A, Morris VK, Allegra CJ, et al. ctDNA applications and integration in colorectal cancer: an NCI Colon and Rectal-Anal Task Forces whitepaper. Nat Rev Clin Oncol 2020;17:757–770. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41571-020-0392-0&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F09%2F06%2F2022.09.03.22279571.atom) 32. 32.Iwaya T, Endo F, Takahashi F, et al. Frequent Tumor Burden Monitoring of Esophageal Squamous Cell Carcinoma With Circulating Tumor DNA Using Individually Designed Digital Polymerase Chain Reaction. Gastroenterology 2021;160:463-+. 33. 33.Yaegashi M, Iwaya T, Sasaki N, et al. Frequent post-operative monitoring of colorectal cancer using individualised ctDNA validated by multiregional molecular profiling. Br J Cancer 2021;124:1556–1565. 34. 34.Sasaki N, Iwaya T, Chiba T, et al. Analysis of mutational and proteomic heterogeneity of gastric cancer suggests an effective pipeline to monitor post-treatment tumor burden using circulating tumor DNA. PLoS One 2020;15:e0239966. 35. 35.Fujisawa R, Iwaya T, Endo F, et al. Early dynamics of circulating tumor DNA predict chemotherapy responses for patients with esophageal cancer. Carcinogenesis 2021;42:1239–1249. 36. 36.Nakanishi H, Sawada T, Kaizaki Y, et al. Significance of gene mutations in the Wnt signaling pathway in traditional serrated adenomas of the colon and rectum. PLoS One 2020;15:e0229262. 37. 37.Catalogue Of Somatic Mutations In Cancer (COSMIC). [https://cancer.sanger.ac.uk/cosmic](https://cancer.sanger.ac.uk/cosmic) 2021. 38. 38.Parkinson CA, Gale D, Piskorz AM, et al. Exploratory Analysis of TP53 Mutations in Circulating Tumour DNA as Biomarkers of Treatment Response for Patients with Relapsed High-Grade Serous Ovarian Carcinoma: A Retrospective Study. PLoS Med 2016;13:e1002198. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1371/journal.pmed.1002198&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F09%2F06%2F2022.09.03.22279571.atom) 39. 39.Kobayashi S, Nakamura Y, Taniguchi H, et al. Impact of Preoperative Circulating Tumor DNA Status on Survival Outcomes After Hepatectomy for Resectable Colorectal Liver Metastases. Ann Surg Oncol 2021;28:4744–4755. 40. 40.de González AB, Darby S. Risk of cancer from diagnostic X-rays: estimates for the UK and 14 other countries. The Lancet 2004;363:345–351. 41. 41.Konishi T, Shimada Y, Hsu M, et al. Association of Preoperative and Postoperative Serum Carcinoembryonic Antigen and Colon Cancer Outcome. JAMA Oncol 2018;4:309–315. 42. 42.Heyries KA. Megapixel digital PCR. Nature Methods 2011;8:649–651. 43. 43.Tie J, Wang Y, Cohen J, et al. Circulating tumor DNA dynamics and recurrence risk in patients undergoing curative intent resection of colorectal cancer liver metastases: A prospective cohort study. PLoS Med 2021;18:e1003620.